Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cell Rep Methods ; 3(5): 100465, 2023 05 22.
Article in English | MEDLINE | ID: mdl-37323577

ABSTRACT

Cellular differentiation requires global changes to DNA methylation (DNAme), where it functions to regulate transcription factor, chromatin remodeling activity, and genome interpretation. Here, we describe a simple DNAme engineering approach in pluripotent stem cells (PSCs) that stably extends DNAme across target CpG islands (CGIs). Integration of synthetic CpG-free single-stranded DNA (ssDNA) induces a target CpG island methylation response (CIMR) in multiple PSC lines, Nt2d1 embryonal carcinoma cells, and mouse PSCs but not in highly methylated CpG island hypermethylator phenotype (CIMP)+ cancer lines. MLH1 CIMR DNAme spanned the CGI, was precisely maintained through cellular differentiation, suppressed MLH1 expression, and sensitized derived cardiomyocytes and thymic epithelial cells to cisplatin. Guidelines for CIMR editing are provided, and initial CIMR DNAme is characterized at TP53 and ONECUT1 CGIs. Collectively, this resource facilitates CpG island DNAme engineering in pluripotency and the genesis of novel epigenetic models of development and disease.


Subject(s)
DNA Methylation , Neoplasms , Animals , Mice , DNA Methylation/genetics , CpG Islands/genetics , DNA, Single-Stranded/metabolism , Neoplasms/genetics , Epithelial Cells/metabolism
2.
G3 (Bethesda) ; 12(11)2022 11 04.
Article in English | MEDLINE | ID: mdl-35997588

ABSTRACT

MHCY is a second major histocompatibility complex-like gene region in chickens originally identified by the presence of major histocompatibility complex class I-like and class II-like gene sequences. Up to now, the MHCY gene region has been poorly represented in genomic sequence data. A high density of repetitive sequence and multiple members of several gene families prevented the accurate assembly of short-read sequence data for MHCY. Identified here by single-molecule real-time sequencing sequencing of BAC clones for the Gallus gallus Red Jungle Fowl reference genome are 107 MHCY region genes (45 major histocompatibility complex class I-like, 41 c-type-lectin-like, 8 major histocompatibility complex class IIß, 8 LENG9-like, 4 zinc finger protein loci, and a single only zinc finger-like locus) located amid hundreds of retroelements within 4 contigs representing the region. Sequences obtained for nearby ribosomal RNA genes have allowed MHCY to be precisely mapped with respect to the nucleolar organizer region. Gene sequences provide insights into the unusual structure of the MHCY class I molecules. The MHCY class I loci are polymorphic and group into 22 types based on predicted amino acid sequences. Some MHCY class I loci are full-length major histocompatibility complex class I genes. Others with altered gene structure are considered gene candidates. The amino acid side chains at many of the polymorphic positions in MHCY class I are directed away rather than into the antigen-binding groove as is typical of peptide-binding major histocompatibility complex class I molecules. Identical and nearly identical blocks of genomic sequence contribute to the observed multiplicity of identical MHCY genes and the large size (>639 kb) of the Red Jungle Fowl MHCY haplotype. Multiple points of hybridization observed in fluorescence in situ hybridization suggest that the Red Jungle Fowl MHCY haplotype is made up of linked, but physically separated genomic segments. The unusual gene content, the evidence of highly similar duplicated segments, and additional evidence of variation in haplotype size distinguish polymorphic MHCY from classical polymorphic major histocompatibility complex regions.


Subject(s)
Chickens , Genes, MHC Class I , Animals , Chickens/genetics , Haplotypes , DNA Transposable Elements , In Situ Hybridization, Fluorescence , Lectins, C-Type/genetics
3.
Nat Commun ; 12(1): 805, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33547295

ABSTRACT

Efforts to improve the prognosis of steroid-resistant gut acute graft-versus-host-disease (SR-Gut-aGVHD) have suffered from poor understanding of its pathogenesis. Here we show that the pathogenesis of SR-Gut-aGVHD is associated with reduction of IFN-γ+ Th/Tc1 cells and preferential expansion of IL-17-IL-22+ Th/Tc22 cells. The IL-22 from Th/Tc22 cells causes dysbiosis in a Reg3γ-dependent manner. Transplantation of IFN-γ-deficient donor CD8+ T cells in the absence of CD4+ T cells produces a phenocopy of SR-Gut-aGVHD. IFN-γ deficiency in donor CD8+ T cells also leads to a PD-1-dependent depletion of intestinal protective CX3CR1hi mononuclear phagocytes (MNP), which also augments expansion of Tc22 cells. Supporting the dual regulation, simultaneous dysbiosis induction and depletion of CX3CR1hi MNP results in full-blown Gut-aGVHD. Our results thus provide insights into SR-Gut-aGVHD pathogenesis and suggest the potential efficacy of IL-22 antagonists and IFN-γ agonists in SR-Gut-aGVHD therapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dysbiosis/immunology , Graft vs Host Disease/immunology , Interferon-gamma/immunology , Interleukins/immunology , Phagocytes/immunology , Animals , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/transplantation , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/immunology , Disease Models, Animal , Dysbiosis/genetics , Dysbiosis/microbiology , Dysbiosis/pathology , Gastrointestinal Microbiome/immunology , Gene Expression Regulation , Graft vs Host Disease/genetics , Graft vs Host Disease/microbiology , Graft vs Host Disease/pathology , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interleukin-17/deficiency , Interleukin-17/genetics , Interleukin-17/immunology , Interleukins/genetics , Intestines/immunology , Intestines/microbiology , Intestines/pathology , Lymphocyte Depletion , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Pancreatitis-Associated Proteins/genetics , Pancreatitis-Associated Proteins/immunology , Phagocytes/cytology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Signal Transduction , T-Lymphocytes, Helper-Inducer , T-Lymphocytes, Regulatory , Whole-Body Irradiation , Interleukin-22
4.
Nat Commun ; 11(1): 6121, 2020 11 30.
Article in English | MEDLINE | ID: mdl-33257686

ABSTRACT

Modified Vaccinia Ankara (MVA) is a highly attenuated poxvirus vector that is widely used to develop vaccines for infectious diseases and cancer. We demonstrate the construction of a vaccine platform based on a unique three-plasmid system to efficiently generate recombinant MVA vectors from chemically synthesized DNA. In response to the ongoing global pandemic caused by SARS coronavirus-2 (SARS-CoV-2), we use this vaccine platform to rapidly produce fully synthetic MVA (sMVA) vectors co-expressing SARS-CoV-2 spike and nucleocapsid antigens, two immunodominant antigens implicated in protective immunity. We show that mice immunized with these sMVA vectors develop robust SARS-CoV-2 antigen-specific humoral and cellular immune responses, including potent neutralizing antibodies. These results demonstrate the potential of a vaccine platform based on synthetic DNA to efficiently generate recombinant MVA vectors and to rapidly develop a multi-antigenic poxvirus-based SARS-CoV-2 vaccine candidate.


Subject(s)
COVID-19 Vaccines/immunology , Coronavirus Nucleocapsid Proteins/immunology , Spike Glycoprotein, Coronavirus/immunology , Vaccines, Synthetic/immunology , Adaptive Immunity , Animals , Antibodies, Neutralizing , Antibodies, Viral/immunology , Antigens, Viral/immunology , Genetic Vectors/immunology , Humans , Immunity, Cellular , Mice , Phosphoproteins/immunology , SARS-CoV-2/immunology , Vaccines, Attenuated/immunology , Vaccinia virus/immunology , Viral Vaccines/immunology
5.
bioRxiv ; 2020 Jul 02.
Article in English | MEDLINE | ID: mdl-32637957

ABSTRACT

Modified Vaccinia Ankara (MVA) is a highly attenuated poxvirus vector that is widely used to develop vaccines for infectious diseases and cancer. We developed a novel vaccine platform based on a unique three-plasmid system to efficiently generate recombinant MVA vectors from chemically synthesized DNA. In response to the ongoing global pandemic caused by SARS coronavirus-2 (SARS-CoV-2), we used this novel vaccine platform to rapidly produce fully synthetic MVA (sMVA) vectors co-expressing SARS-CoV-2 spike and nucleocapsid antigens, two immunodominant antigens implicated in protective immunity. Mice immunized with these sMVA vectors developed robust SARS-CoV-2 antigen-specific humoral and cellular immune responses, including potent neutralizing antibodies. These results demonstrate the potential of a novel vaccine platform based on synthetic DNA to efficiently generate recombinant MVA vectors and to rapidly develop a multi-antigenic poxvirus-based SARS-CoV-2 vaccine candidate.

7.
Neurosci Lett ; 590: 35-9, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25636690

ABSTRACT

Transcriptomic studies of murine D3 embryonic stem (ES) cells deficient in the purinergic biosynthetic function hypoxanthine guanine phosphoribosyltransferase (HPRT) and undergoing dopaminergic neuronal differentiation has demonstrated a marked shift from neuronal to glial gene expression and aberrant expression of multiple genes also known to be aberrantly expressed in Alzheimer's and other CNS disorders. Such genetic dysregulations may indicate some shared pathogenic metabolic mechanisms in diverse CNS diseases.


Subject(s)
Alzheimer Disease/genetics , Hypoxanthine Phosphoribosyltransferase/deficiency , Lesch-Nyhan Syndrome/genetics , Purines/biosynthesis , Transcriptome , Alzheimer Disease/metabolism , Animals , Cell Differentiation , Cell Line , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Lesch-Nyhan Syndrome/metabolism , Mice , Neuroglia/cytology , Neuroglia/metabolism
8.
Cell Rep ; 10(7): 1158-72, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25704818

ABSTRACT

Neural stem cells (NSCs) continuously produce new neurons within the adult mammalian hippocampus. NSCs are typically quiescent but activated to self-renew or differentiate into neural progenitor cells. The molecular mechanisms of NSC activation remain poorly understood. Here, we show that adult hippocampal NSCs express vascular endothelial growth factor receptor (VEGFR) 3 and its ligand VEGF-C, which activates quiescent NSCs to enter the cell cycle and generate progenitor cells. Hippocampal NSC activation and neurogenesis are impaired by conditional deletion of Vegfr3 in NSCs. Functionally, this is associated with compromised NSC activation in response to VEGF-C and physical activity. In NSCs derived from human embryonic stem cells (hESCs), VEGF-C/VEGFR3 mediates intracellular activation of AKT and ERK pathways that control cell fate and proliferation. These findings identify VEGF-C/VEGFR3 signaling as a specific regulator of NSC activation and neurogenesis in mammals.


Subject(s)
Neural Stem Cells/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Hippocampus/metabolism , Humans , Mice , Mice, Inbred C57BL , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Signal Transduction , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor C/pharmacology , Vascular Endothelial Growth Factor Receptor-3/genetics
9.
PLoS One ; 8(10): e74967, 2013.
Article in English | MEDLINE | ID: mdl-24130677

ABSTRACT

The mechanisms by which mutations of the purinergic housekeeping gene hypoxanthine guanine phosphoribosyltransferase (HPRT) cause the severe neurodevelopmental Lesch Nyhan Disease (LND) are poorly understood. The best recognized neural consequences of HPRT deficiency are defective basal ganglia expression of the neurotransmitter dopamine (DA) and aberrant DA neuronal function. We have reported that HPRT deficiency leads to dysregulated expression of multiple DA-related developmental functions and cellular signaling defects in a variety of HPRT-deficient cells, including human induced pluripotent stem (iPS) cells. We now describe results of gene expression studies during neuronal differentiation of HPRT-deficient murine ESD3 embryonic stem cells and report that HPRT knockdown causes a marked switch from neuronal to glial gene expression and dysregulates expression of Sox2 and its regulator, genes vital for stem cell pluripotency and for the neuronal/glial cell fate decision. In addition, HPRT deficiency dysregulates many cellular functions controlling cell cycle and proliferation mechanisms, RNA metabolism, DNA replication and repair, replication stress, lysosome function, membrane trafficking, signaling pathway for platelet activation (SPPA) multiple neurotransmission systems and sphingolipid, sulfur and glycan metabolism. We propose that the neural aberrations of HPRT deficiency result from combinatorial effects of these multi-system metabolic errors. Since some of these aberrations are also found in forms of Alzheimer's and Huntington's disease, we predict that some of these systems defects play similar neuropathogenic roles in diverse neurodevelopmental and neurodegenerative diseases in common and may therefore provide new experimental opportunities for clarifying pathogenesis and for devising new potential therapeutic targets in developmental and genetic disease.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/enzymology , Hypoxanthine Phosphoribosyltransferase/metabolism , Neurons/cytology , Animals , Blotting, Western , Cell Differentiation/genetics , Cell Differentiation/physiology , Dopamine/metabolism , Hypoxanthine Phosphoribosyltransferase/genetics , Mice , Mice, Knockout , Neurons/enzymology , Signal Transduction
10.
Proc Natl Acad Sci U S A ; 109(9): 3377-82, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22331909

ABSTRACT

Lesch-Nyhan disease (LND) is an X-linked genetic disorder caused by mutations of the hypoxanthine guanine phosphoribosyltransferase (HPRT) purine biosynthesis gene and characterized by aberrant purine metabolism, deficient basal ganglia dopamine levels, dystonia, and severe neurobehavioral manifestations, including compulsive self-injurious behavior. Although available evidence has identified important roles for purinergic signaling in brain development, the mechanisms linking HPRT deficiency, purinergic pathways, and neural dysfunction of LND are poorly understood. In these studies aimed at characterizing purinergic signaling in HPRT deficiency, we used a lentivirus vector stably expressing an shRNA targeted to the HPRT gene to produce HPRT-deficient human CVB induced pluripotent stem cells and human HUES11 embryonic stem cells. Both CVB and HUES11 cells show >99% HPRT knockdown and demonstrate markedly decreased expression of the purinergic P2Y1 receptor mRNA. In CVB cells, P2Y1 mRNA and protein down-regulation by HPRT knockdown is refractory to activation by the P2Y1 receptor agonist ATP and shows aberrant purinergic signaling, as reflected by marked deficiency of the transcription factor pCREB and constitutive activation of the MAP kinases phospho-ERK1/2. Moreover, HPRT-knockdown CVB cells also demonstrate marked reduction of phosphorylated ß-catenin. These results indicate that the housekeeping gene HPRT regulates purinergic signaling in pluripotent human stem cells, and that this regulation occurs at least partly through aberrant P2Y1-mediated expression and signaling. We propose that such mechanisms may play a role in the neuropathology of HPRT-deficiency LND and may point to potential molecular targets for modulation of this intractable neurological phenotype.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/physiology , Neurogenesis/physiology , Pluripotent Stem Cells/enzymology , Purines/metabolism , Adenosine Triphosphate/pharmacology , Cell Line , Fibroblasts/enzymology , Gene Knockdown Techniques , Genes, Essential , Genetic Vectors/genetics , Glycogen Synthase Kinase 3/physiology , Glycogen Synthase Kinase 3 beta , Humans , Lentivirus/genetics , Lesch-Nyhan Syndrome/enzymology , MAP Kinase Signaling System/physiology , Male , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Purinergic P2Y Receptor Agonists/pharmacology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Receptors, Purinergic P2Y1/genetics , Receptors, Purinergic P2Y1/physiology , beta Catenin/metabolism
11.
PLoS One ; 6(1): e16572, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21305049

ABSTRACT

We have used microarray-based methods of global gene expression together with quantitative PCR and Western blot analysis to identify dysregulation of genes and aberrant cellular processes in human fibroblasts and in SH-SY5Y neuroblastoma cells made HPRT-deficient by transduction with a retrovirus stably expressing an shRNA targeted against HPRT. Analysis of the microarray expression data by Gene ontology (GO) and Gene Set Enrichment Analysis (GSEA) as well as significant pathway analysis by GeneSpring GX10 and Panther Classification System reveal that HPRT deficiency is accompanied by aberrations in a variety of pathways known to regulate neurogenesis or to be implicated in neurodegenerative disease, including the canonical Wnt/ß-catenin and the Alzheimer's disease/presenilin signaling pathways. Dysregulation of the Wnt/ß-catenin pathway is confirmed by Western blot demonstration of cytosolic sequestration of ß-catenin during in vitro differentiation of the SH-SY5Y cells toward the neuronal phenotype. We also demonstrate that two key transcription factor genes known to be regulated by Wnt signaling and to be vital for the generation and function of dopaminergic neurons; i.e., Lmx1a and Engrailed 1, are down-regulated in the HPRT knockdown SH-SY5Y cells. In addition to the Wnt signaling aberration, we found that expression of presenilin-1 shows severely aberrant expression in HPRT-deficient SH-SY5Y cells, reflected by marked deficiency of the 23 kDa C-terminal fragment of presenilin-1 in knockdown cells. Western blot analysis of primary fibroblast cultures from two LND patients also shows dysregulated presenilin-1 expression, including aberrant proteolytic processing of presenilin-1. These demonstrations of dysregulated Wnt signaling and presenilin-1 expression together with impaired expression of dopaminergic transcription factors reveal broad pleitropic neuro-regulatory defects played by HPRT expression and suggest new directions for investigating mechanisms of aberrant neurogenesis and neuropathology in LND and potential new targets for restoration of effective signaling in this neuro-developmental defect.


Subject(s)
Hypoxanthine Phosphoribosyltransferase/deficiency , Neurogenesis/genetics , Presenilin-1/metabolism , Signal Transduction/genetics , Wnt Proteins/metabolism , Cell Line, Tumor , Dopamine , Gene Expression Profiling/methods , Humans , Lesch-Nyhan Syndrome , Transcription Factors
12.
Proteomics ; 7(15): 2624-35, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17610204

ABSTRACT

Oxidative stress is one of the major causes of neuronal cell death in disorders such as perinatal hypoxia and ischemia. Protein phosphorylation is the most significant PTM of proteins and plays an important role in stress-induced signal transduction. Thus, the analysis of alternative protein phosphorylation states which occur during oxidative stress-induced cell death could provide valuable information regarding cell death. In this study, a reference phosphoproteome map of the mouse hippocampal cell line HT22 was constructed based on 125 spots that were identified by MALDI-TOF or LC-ESI-Q-TOF-MS analysis. In addition, proteins of HT22 cells at various stages of oxidative stress-induced cell death were separated by 2-DE and alterations in phosphoproteins were detected by Pro-Q Diamond staining. A total of 17 spots showing significant quantitative changes and seven newly appearing spots were identified after glutamate treatment. Splicing factor 2, peroxiredoxin 2, S100 calcium binding protein A11, and purine nucleoside phosphorylase were identified as up- or down-regulated proteins. CDC25A, caspase-8, and cyp51 protein appeared during oxidative stress-induced cell death. The data in this study from phosphoproteomic analysis provide a valuable resource for the understanding of HT22 cell death mechanisms mediated by oxidative stress.


Subject(s)
Apoptosis/drug effects , Glutamic Acid/pharmacology , Neurons/drug effects , Oxidative Stress/drug effects , Proteome/analysis , Animals , Cell Death/drug effects , Cell Line , Cell Survival/drug effects , Edetic Acid/pharmacology , Egtazic Acid/pharmacology , Hippocampus/cytology , Mass Spectrometry , Mice , Neurons/metabolism , Phosphoproteins/analysis , Phosphoproteins/metabolism , Phosphorylation , Proteomics/methods , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...